Abstract

Aims

To perform a thorough and updated systematic review of randomized clinical trials comparing tirofiban vs. placebo or vs. abciximab.

Methods and results

We searched for randomized trials comparing tirofiban vs. placebo or any active control. Odds ratios (OR) were computed from individual studies and pooled with random-effect methods. Thirty-one studies were identified involving 20 006 patients (12 874 comparing tirofiban vs. heparin plus placebo or bivalirudin alone, and 7132 vs. abciximab). When compared with placebo, tirofiban was associated at 30 days with a significant reduction in mortality [OR = 0.68 (0.54–0.86); P = 0.001] and death or myocardial infarction (MI) [OR = 0.69 (0.58–0.81); P < 0.001]. The treatment benefit persisted at follow-up but came at an increased risk of minor bleedings [OR = 1.42 (1.13, 1.79), P = 0.002] or thrombocytopenia. When compared with abciximab, mortality at 30 days did not differ [OR = 0.90 (0.53, 1.54); P = 0.70], but in the overall group tirofiban trended to increase the composite of death or MI [OR = 1.18 (0.96, 1.45); P = 0.11]. No such trend persisted at medium-term follow-up or when appraising studies testing tirofiban at 25 µg/kg bolus regimen.

Conclusion

Tirofiban administration reduces mortality, the composite of death or MI and increases minor bleedings when compared with placebo. An early ischaemic hazard disfavouring tirofiban was noted when compared with abciximab in studies based on 10 but not 25 µg/kg tirofiban bolus regimen.

Introduction

Tirofiban is a small molecule, non-peptide tyrosine derivative which belongs to the class of glycoprotein (GP) IIb/IIIa inhibitors (GPI).1,2 By preventing the binding of fibrinogen and von Willebrand factor to the GP IIb/IIIa receptor on the surface of the platelet, GPIs are currently regarded as the most potent inhibitors of platelet aggregation.1,2

Though similar to abciximab in that it has a high affinity for the GP IIb/IIIa receptor, tirofiban dissociates from the GP IIb/IIIa receptor more rapidly than abciximab.1,2 Its anti-aggretory effects reverse within hours after the completion of the infusion, whereas abciximab binds near irreversibly to the receptor resulting in a considerably longer effect.1,3 Additionally, tirofiban does not inhibit other β3 integrins, such as the vitronectin receptor, at the surface of vascular cells or the activated MAC-1 receptor on leucocytes,4 which have been traditionally regarded as crucial targets to explain abciximab effects on microcirculation.5

Even more importantly, different dosing regimens of tirofiban have been developed over time based on the clinical setting and the timing of percutaneous coronary intervention (PCI) which has resulted in mixed results in clinical trials when compared with either placebo or abciximab.3,6–10 Thus, uncertainty on the role of tirofiban still largely persists in current clinical practice.

Systematic reviews employing meta-analytic techniques provide quantitative and objective means to pool and assess available clinical evidence, emphasizing internal validity and homogeneity, while affording increased statistical power for hypothesis testing. The aim of this study was thus to perform a thorough and updated systematic review of randomized clinical trials comparing tirofiban vs. placebo or vs. abciximab in patients undergoing treatment for various coronary artery disease (CAD) conditions, with specific emphasis on the role of front-loaded tirofiban regimen and timing of intervention.

Methods

Search strategy

Two expert cardiologists (M.V., M.T.) independently and systematically searched BioMedCentral, CENTRAL, Clinicaltrials.gov, EMBASE, and PubMed for randomized trials comparing tirofiban vs. placebo or any active control in patients with acute coronary syndromes and/or undergoing PCI (updated October 2008), with divergences resolved after consensus.11 EMBASE and PubMed were searched with explode features according to the following strategy: ‘(abciximab[all] OR tirofiban[all] OR (glycoprotein[all] AND (iib/iiia OR iibiiia) AND inhibitor*[all])) AND coronary AND (clinical trial*[all] OR random*[all])’.12

Articles published in languages other than English or Italian (the native languages of the authors) were systematically searched in multiple online databases, international conference proceedings, references (backward snowballing) or cross-quotations (forward snowballing) from included studies and pertinent available quantitative reviews, and queries to international experts. References were systematically scanned to retrieve additional studies. No language restriction was enforced.

Selection criteria

Shortlisted studies were retrieved as full articles and appraised by three unblinded reviewers independently (M.V., G.B.Z., M.T.), with divergences resolved after consensus, according to the following inclusion criteria: (i) randomized treatment allocation and (ii) comparison of tirofiban vs. placebo or active. Exclusion criteria were: (i) duplicate reports failing to report additional or extended clinical outcomes, (ii) lack of outcome data beyond hospitalization, and (iii) equivocal (i.e. no clear information on modalities for allocating patients to tirofiban vs. placebo or active treatment) or non-random treatment allocation.

Data abstraction and validity assessment

Three reviewers independently abstracted data, with divergences resolved after consensus. In case of incomplete or unclear data, authors were contacted where possible. The co-primary endpoints of the analysis were the 30 days and long-term mortality rates. The incidence of major adverse cardiovascular events (MACE), including the composite of death, myocardial infarction (MI), or urgent revascularization, death or MI, as well as major and minor bleeding [according to the Thrombolysis In Myocardial Infarction (TIMI) criteria]13 and thrombocytopenia were also appraised. We pre hoc stratified studies according to the type of control, dosage/timing of tirofiban administration, and type of concomitant oral anti-platelet therapy. Thus, studies where more than one dosage or timing of intervention were tested vs. placebo or active control have been split up into the most suitable pre-specified study categories. Additional pertinent data for baseline and procedural characteristics were abstracted, including type of PCI. Study validity and risk of bias were appraised according to The Cochrane Collaboration methods, i.e. separately appraising means for generating the randomization sequence, allocation concealment, blinding, concurrent treatment, data completion, definitions, outcome reporting, other potential source of bias, and overall risk of bias.11

Data analysis and synthesis

Odds ratios (OR) were computed from individual studies and pooled according to DerSimonian-Laird random-effect methods (with 95% confidence intervals) using RevMan 4.2 (The Cochrane Collaboration, Købehavn, Denmark). Inconsistency was appraised by means of I2. Specifically, I2 < 25% suggests mild, statistical inconsistency, whereas I2 values in the 25–50% range and in the 75−100% represent, respectively, moderate and extensive inconsistency. Statistical heterogeneity was appraised with χ2 tests, with P-values less than 0.10 suggesting underlying heterogeneity. Small study bias and/or publication bias (i.e. the likelihood of small yet nominally significant studies being selectively published in the literature) were appraised by means of visual inspection of funnel plots and Peters test.14 Random-effect meta-regression was also performed to explore moderators and/or predictors of changes in log-transformed OR, by means of a weighted-least-square inverse-variance weighted method with SPSS 11.0 (SPSS, Chicago, IL, USA). Unadjusted P-values are reported throughout, with hypothesis testing set at the two-tailed 0.05 level.

According to absolute risk reduction or increment obtained with random-effect risk differences computed at 30-day follow-up, we calculated the number needed to treat (NNT) to prevent one event, whereas for the safety endpoints, we calculated the number needed to harm (NNH) to determine one adverse event.

Results

Search results and study selection

Database searches retrieved 1952 citations (Figure 1). Shortlisted citations were retrieved and checked at the title/abstract level excluding 1894 papers. Complete articles for the remaining 58 studies were checked for compliance to inclusion/exclusion criteria. Reasons for further exclusion included non-experimental design, use of historical controls, duplicate reporting, unconventional study drug regimen, early vs. late study drug administration design, or failure to report/provide upon request clinical data. We finally identified 31 eligible trials of which 22 were controlled with placebo6–8,15–35 (Table 1), eight with abciximab,9,10,36–41 and one with both agents42 (Table 2).

Figure 1

Flow diagram of the systematic literature search indicating the inclusion and exclusion process of studies.

Table 1

Main characteristics of the placebo-controlled trials

StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
3T/2R352006–2008263ASA and/or clopidogrel poor respondersBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH or bivalirudin, and clopidogrelPeriprocedural MI defined as troponin >3× ULN within 48 h30 Days
ADVANCE252002–2003202High-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidine or clopidogrelDeath, MI, TVR, or bailout tirofiban6 Months
ELISA 2292002–2005328NSTEACSBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, LWMH, clopdidogrelEnzymatic infarct size (LDHQ 48)30 Days
Ercan et al.22Not reported57NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, clopidogrelLevels of C-reactive protein at 48 h30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelPlatelet aggregation inhibitionHospital stay
Fu et al.302005–2007150STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNot reported30 Days
Ivandic et al.312004–2006100NSTEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelInfarct size based on troponin T elevation6 Months
Juergens et al.18Not reported894Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHTIMI bleedings30 Days
Kereiakes et al.15Not reported44Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHPlatelet aggregation inhibitionHospital stay
Kim et al.262001–2002160NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH or dalteparin,Not reported6 Months
Kurowski et al.272000–200350Stable or marker-negative unstable angina undergoing SVG stentingBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMyocardial necrosis as evidenced by an increase in the cTnT above the ULN within 72 h21 Months
NAPLES342005–2008335Elective PCI in diabeticsBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNACE30 Days
Okmen et al.21Not reported83NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHInfarct size based on CK-MB >2× ULN10 Months
Okmen et al.24Not reported119Stable or unstable angina undergoing PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, ticlopidine, UFHInfarct size based on CK-MB >2× ULN21 Months
On-TIME 2 open-label study332004–2006414STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelNone30 Days
On-TIME 2332006–2007984STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelResidual ST-segment deviation at ECG30 Days
Ozkan et al.281999–200447Stable or marker-negative unstable angina undergoing SVG stentingBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, enoxaparin, and clopidogrelNot reported30 Days
PRISM81994–19963232NSTEACSBolus: 0.6 µg/kg/min×30 min; infusion: 0.15 µg/kg/minUpstreamASA, UFH in placebo group onlyDeath, MI, refractory ischemia at 48 h30 Days
PRISM-PLUS71994–19961570NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHDeath, MI, refractory ischemia at 7 days6 Months
RESTORE619952212NSTEACS or STEMI undergoing POBA or DCABolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHDeath, MI, TVR, or bailout stenting6 Months
SASTRE232000–2002144STEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, alteplaseTIMI flow grade 3 in the IRA at 90′30 Days
Shen et al.322005–2006115STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMACE at 30 days6 Months
TETAMI19,201999–20021224STEMI ineligible to lysisBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, enoxaparin or UFHDeath, MI, recurrent angina30 Days
TOPSTAR17Not reported96Stable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTroponin elevation within 48 h9 Months
StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
3T/2R352006–2008263ASA and/or clopidogrel poor respondersBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH or bivalirudin, and clopidogrelPeriprocedural MI defined as troponin >3× ULN within 48 h30 Days
ADVANCE252002–2003202High-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidine or clopidogrelDeath, MI, TVR, or bailout tirofiban6 Months
ELISA 2292002–2005328NSTEACSBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, LWMH, clopdidogrelEnzymatic infarct size (LDHQ 48)30 Days
Ercan et al.22Not reported57NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, clopidogrelLevels of C-reactive protein at 48 h30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelPlatelet aggregation inhibitionHospital stay
Fu et al.302005–2007150STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNot reported30 Days
Ivandic et al.312004–2006100NSTEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelInfarct size based on troponin T elevation6 Months
Juergens et al.18Not reported894Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHTIMI bleedings30 Days
Kereiakes et al.15Not reported44Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHPlatelet aggregation inhibitionHospital stay
Kim et al.262001–2002160NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH or dalteparin,Not reported6 Months
Kurowski et al.272000–200350Stable or marker-negative unstable angina undergoing SVG stentingBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMyocardial necrosis as evidenced by an increase in the cTnT above the ULN within 72 h21 Months
NAPLES342005–2008335Elective PCI in diabeticsBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNACE30 Days
Okmen et al.21Not reported83NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHInfarct size based on CK-MB >2× ULN10 Months
Okmen et al.24Not reported119Stable or unstable angina undergoing PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, ticlopidine, UFHInfarct size based on CK-MB >2× ULN21 Months
On-TIME 2 open-label study332004–2006414STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelNone30 Days
On-TIME 2332006–2007984STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelResidual ST-segment deviation at ECG30 Days
Ozkan et al.281999–200447Stable or marker-negative unstable angina undergoing SVG stentingBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, enoxaparin, and clopidogrelNot reported30 Days
PRISM81994–19963232NSTEACSBolus: 0.6 µg/kg/min×30 min; infusion: 0.15 µg/kg/minUpstreamASA, UFH in placebo group onlyDeath, MI, refractory ischemia at 48 h30 Days
PRISM-PLUS71994–19961570NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHDeath, MI, refractory ischemia at 7 days6 Months
RESTORE619952212NSTEACS or STEMI undergoing POBA or DCABolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHDeath, MI, TVR, or bailout stenting6 Months
SASTRE232000–2002144STEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, alteplaseTIMI flow grade 3 in the IRA at 90′30 Days
Shen et al.322005–2006115STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMACE at 30 days6 Months
TETAMI19,201999–20021224STEMI ineligible to lysisBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, enoxaparin or UFHDeath, MI, recurrent angina30 Days
TOPSTAR17Not reported96Stable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTroponin elevation within 48 h9 Months

ASA, aspirin; LWMH, low-weight molecular heparin; LDH, lactate dehydrogenase; NSTEACS, non-ST-segment elevation acute coronary syndromes; NSTEMI, non-ST-segment elevation myocardial infarction; MI, myocardial infarction; LDHQ48, area under the lactate dehydrogenase release over 48 h curve; POBA, balloon angioplasty; DCA, directional coronary atherectomy; STEMI, ST-segment elevation myocardial infarction; ULN, upper limit of normal; PCI, percutaneous coronary intervention; SVG, saphenous vein graft; IRA, infarct-related artery; MACE, major adverse cardiovascular events; NACE, net adverse cardiovascular events; UFH, unfractionated heparin; TVR, target vessel revascularization.

Table 1

Main characteristics of the placebo-controlled trials

StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
3T/2R352006–2008263ASA and/or clopidogrel poor respondersBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH or bivalirudin, and clopidogrelPeriprocedural MI defined as troponin >3× ULN within 48 h30 Days
ADVANCE252002–2003202High-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidine or clopidogrelDeath, MI, TVR, or bailout tirofiban6 Months
ELISA 2292002–2005328NSTEACSBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, LWMH, clopdidogrelEnzymatic infarct size (LDHQ 48)30 Days
Ercan et al.22Not reported57NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, clopidogrelLevels of C-reactive protein at 48 h30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelPlatelet aggregation inhibitionHospital stay
Fu et al.302005–2007150STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNot reported30 Days
Ivandic et al.312004–2006100NSTEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelInfarct size based on troponin T elevation6 Months
Juergens et al.18Not reported894Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHTIMI bleedings30 Days
Kereiakes et al.15Not reported44Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHPlatelet aggregation inhibitionHospital stay
Kim et al.262001–2002160NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH or dalteparin,Not reported6 Months
Kurowski et al.272000–200350Stable or marker-negative unstable angina undergoing SVG stentingBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMyocardial necrosis as evidenced by an increase in the cTnT above the ULN within 72 h21 Months
NAPLES342005–2008335Elective PCI in diabeticsBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNACE30 Days
Okmen et al.21Not reported83NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHInfarct size based on CK-MB >2× ULN10 Months
Okmen et al.24Not reported119Stable or unstable angina undergoing PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, ticlopidine, UFHInfarct size based on CK-MB >2× ULN21 Months
On-TIME 2 open-label study332004–2006414STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelNone30 Days
On-TIME 2332006–2007984STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelResidual ST-segment deviation at ECG30 Days
Ozkan et al.281999–200447Stable or marker-negative unstable angina undergoing SVG stentingBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, enoxaparin, and clopidogrelNot reported30 Days
PRISM81994–19963232NSTEACSBolus: 0.6 µg/kg/min×30 min; infusion: 0.15 µg/kg/minUpstreamASA, UFH in placebo group onlyDeath, MI, refractory ischemia at 48 h30 Days
PRISM-PLUS71994–19961570NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHDeath, MI, refractory ischemia at 7 days6 Months
RESTORE619952212NSTEACS or STEMI undergoing POBA or DCABolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHDeath, MI, TVR, or bailout stenting6 Months
SASTRE232000–2002144STEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, alteplaseTIMI flow grade 3 in the IRA at 90′30 Days
Shen et al.322005–2006115STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMACE at 30 days6 Months
TETAMI19,201999–20021224STEMI ineligible to lysisBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, enoxaparin or UFHDeath, MI, recurrent angina30 Days
TOPSTAR17Not reported96Stable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTroponin elevation within 48 h9 Months
StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
3T/2R352006–2008263ASA and/or clopidogrel poor respondersBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH or bivalirudin, and clopidogrelPeriprocedural MI defined as troponin >3× ULN within 48 h30 Days
ADVANCE252002–2003202High-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidine or clopidogrelDeath, MI, TVR, or bailout tirofiban6 Months
ELISA 2292002–2005328NSTEACSBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, LWMH, clopdidogrelEnzymatic infarct size (LDHQ 48)30 Days
Ercan et al.22Not reported57NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, clopidogrelLevels of C-reactive protein at 48 h30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelPlatelet aggregation inhibitionHospital stay
Fu et al.302005–2007150STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNot reported30 Days
Ivandic et al.312004–2006100NSTEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelInfarct size based on troponin T elevation6 Months
Juergens et al.18Not reported894Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHTIMI bleedings30 Days
Kereiakes et al.15Not reported44Elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHPlatelet aggregation inhibitionHospital stay
Kim et al.262001–2002160NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH or dalteparin,Not reported6 Months
Kurowski et al.272000–200350Stable or marker-negative unstable angina undergoing SVG stentingBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMyocardial necrosis as evidenced by an increase in the cTnT above the ULN within 72 h21 Months
NAPLES342005–2008335Elective PCI in diabeticsBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelNACE30 Days
Okmen et al.21Not reported83NSTEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHInfarct size based on CK-MB >2× ULN10 Months
Okmen et al.24Not reported119Stable or unstable angina undergoing PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, ticlopidine, UFHInfarct size based on CK-MB >2× ULN21 Months
On-TIME 2 open-label study332004–2006414STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelNone30 Days
On-TIME 2332006–2007984STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelResidual ST-segment deviation at ECG30 Days
Ozkan et al.281999–200447Stable or marker-negative unstable angina undergoing SVG stentingBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, enoxaparin, and clopidogrelNot reported30 Days
PRISM81994–19963232NSTEACSBolus: 0.6 µg/kg/min×30 min; infusion: 0.15 µg/kg/minUpstreamASA, UFH in placebo group onlyDeath, MI, refractory ischemia at 48 h30 Days
PRISM-PLUS71994–19961570NSTEACSBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFHDeath, MI, refractory ischemia at 7 days6 Months
RESTORE619952212NSTEACS or STEMI undergoing POBA or DCABolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHDeath, MI, TVR, or bailout stenting6 Months
SASTRE232000–2002144STEMIBolus: 0.4 µg/kg/min×30 min; infusion: 0.1 µg/kg/minUpstreamASA, UFH, alteplaseTIMI flow grade 3 in the IRA at 90′30 Days
Shen et al.322005–2006115STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, UFH, clopidogrelMACE at 30 days6 Months
TETAMI19,201999–20021224STEMI ineligible to lysisBolus: 10 µg/kg; infusion: 0.15 µg/kg/minUpstreamASA, enoxaparin or UFHDeath, MI, recurrent angina30 Days
TOPSTAR17Not reported96Stable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTroponin elevation within 48 h9 Months

ASA, aspirin; LWMH, low-weight molecular heparin; LDH, lactate dehydrogenase; NSTEACS, non-ST-segment elevation acute coronary syndromes; NSTEMI, non-ST-segment elevation myocardial infarction; MI, myocardial infarction; LDHQ48, area under the lactate dehydrogenase release over 48 h curve; POBA, balloon angioplasty; DCA, directional coronary atherectomy; STEMI, ST-segment elevation myocardial infarction; ULN, upper limit of normal; PCI, percutaneous coronary intervention; SVG, saphenous vein graft; IRA, infarct-related artery; MACE, major adverse cardiovascular events; NACE, net adverse cardiovascular events; UFH, unfractionated heparin; TVR, target vessel revascularization.

Table 2

Main characteristics of the abciximab-controlled trials

StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
Danzi et al.372002100STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHInfarct-zone wall motion score index at 30 days30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ClopidogrelPlatelet aggregation inhibitionHospital stay
EVEREST362003–200461NSTEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTIMI myocardial perfusion rate30 Days
FATA382004–2007692STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH≥70% STR at 90′30 Days
MULTISTRATEGY102004–2007744STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrel≥50% STR at 90′8 Months
Neumann et al.40Not reported40Stable or unstable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidineInhibition of platelet aggregation after 2 h of infusion30 Days
STRATEGY412003–2004175STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, stroke, binary restenosis36 Months
TARGET91999–20005308Urgent and elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR at 30 days12 Months
TENACITY432004–2005383Medium to high-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR30 Days
StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
Danzi et al.372002100STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHInfarct-zone wall motion score index at 30 days30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ClopidogrelPlatelet aggregation inhibitionHospital stay
EVEREST362003–200461NSTEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTIMI myocardial perfusion rate30 Days
FATA382004–2007692STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH≥70% STR at 90′30 Days
MULTISTRATEGY102004–2007744STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrel≥50% STR at 90′8 Months
Neumann et al.40Not reported40Stable or unstable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidineInhibition of platelet aggregation after 2 h of infusion30 Days
STRATEGY412003–2004175STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, stroke, binary restenosis36 Months
TARGET91999–20005308Urgent and elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR at 30 days12 Months
TENACITY432004–2005383Medium to high-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR30 Days

ASA, aspirin; UFH, unfractionated heparin; NSTEMI, non-ST-segment elevation myocardial infarction; MI, myocardial infarction; PCI, percutaneous coronary intervention; MI, myocardial infarction; TVR, target vessel revascularization; STR, ST-segment resolution.

Table 2

Main characteristics of the abciximab-controlled trials

StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
Danzi et al.372002100STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHInfarct-zone wall motion score index at 30 days30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ClopidogrelPlatelet aggregation inhibitionHospital stay
EVEREST362003–200461NSTEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTIMI myocardial perfusion rate30 Days
FATA382004–2007692STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH≥70% STR at 90′30 Days
MULTISTRATEGY102004–2007744STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrel≥50% STR at 90′8 Months
Neumann et al.40Not reported40Stable or unstable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidineInhibition of platelet aggregation after 2 h of infusion30 Days
STRATEGY412003–2004175STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, stroke, binary restenosis36 Months
TARGET91999–20005308Urgent and elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR at 30 days12 Months
TENACITY432004–2005383Medium to high-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR30 Days
StudyTrial periodNo. randomizedPatient populationTirofiban doseSettingConcomitant anti-thrombotics1° EndpointFollow-up
Danzi et al.372002100STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFHInfarct-zone wall motion score index at 30 days30 Days
Ernst et al.422002–200360STEMIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ClopidogrelPlatelet aggregation inhibitionHospital stay
EVEREST362003–200461NSTEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelTIMI myocardial perfusion rate30 Days
FATA382004–2007692STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH≥70% STR at 90′30 Days
MULTISTRATEGY102004–2007744STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrel≥50% STR at 90′8 Months
Neumann et al.40Not reported40Stable or unstable anginaBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, ticlopidineInhibition of platelet aggregation after 2 h of infusion30 Days
STRATEGY412003–2004175STEMIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, stroke, binary restenosis36 Months
TARGET91999–20005308Urgent and elective PCIBolus: 10 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR at 30 days12 Months
TENACITY432004–2005383Medium to high-risk PCIBolus: 25 µg/kg; infusion: 0.15 µg/kg/minDownstreamASA, UFH, clopidogrelDeath, MI, urgent TVR30 Days

ASA, aspirin; UFH, unfractionated heparin; NSTEMI, non-ST-segment elevation myocardial infarction; MI, myocardial infarction; PCI, percutaneous coronary intervention; MI, myocardial infarction; TVR, target vessel revascularization; STR, ST-segment resolution.

Study and patient characteristics

The 31 studies included in the final analysis 20 006 randomized patients (average follow-up 5 months), 12 874 vs. placebo and 7132 vs. abciximab. In seven placebo-controlled trials,7,8,21–23,26,28 mainly recruiting patients with confirmed or suspected non-ST-segment elevation acute coronary syndrome (NSTEACS) or ST-elevation myocardial infarction (STEMI), tirofiban was co-administered together with unfractioned heparin (UFH) upon presentation (upstream) as a 0.4 µg/kg 30 min bolus regimen followed by 0.1 µg/kg/min infusion apart from PRISM study,8 where tirofiban was given without UFH at a 0.6 µg/kg 30 min bolus regimen followed by 0.15 µg/kg/min infusion.

In five placebo-controlled studies,19,20,27,29,31,32 tirofiban was administered upstream in NSTEACS, STEMI, or stable CAD patients at 10 µg/kg 3 min bolus regimen and 0.15 µg/kg/min infusion, whereas in 12 studies, eight placebo-controlled,6,15,17,18,24,30,32,34 two abciximab-controlled,9,39,40 one controlled with both agents,42 and one with bivalirudin,34 tirofiban was given at 10 µg/kg 3 min bolus regimen and 0.15 µg/kg/min infusion prior PCI (downstream) in patients with stable or unstable CAD. Finally, in 4076 patients tirofiban was administered downstream at high bolus dose (25 µg/kg 3 min bolus regimen and 0.15 µg/kg/min infusion) and controlled with placebo (n = 1863)25,33,35,42 or abciximab (n = 2213).10,36–38,41–43 Study quality and risk of bias were variable, reflecting heterogeneity in setting, sample size, and study design (phase III vs. IV) and is detailed in Table 3.

Table 3

Risk of bias assessment

StudyAdequate sequence generationAllocation concealment usedBlindingConcurrent therapies similarIncomplete outcome data addressedUniform and explicit outcome definitionsFree of selective outcome reportingFree of other biasOverall risk of bias
3T/2R35Yes (computer generated)Yes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
ADVANCE25Yes (computer generated)Yes (external personnel)Yes (patients, caring physicians, and outcome assessors)YesNoYesYesYesLow
Danzi et al.37UnclearUnclearNoYesYesNoNoYesModerate
ELISA 229UnclearUnclearYes (outcome assessors)NoYesYesYesYesModerate
Ercan et al.22UnclearUnclearNoNoYesYesYesYesModerate
Ernst et al.42Yes (computer generated)UnclearNoYesNoYesYesYesModerate
EVEREST36UnclearUnclearNoYesYesNoNoYesModerate
FATA38UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
Fu et al.30UnclearUnclearNoYesNoNoYesYesModerate
Ivandic et al.31UnclearUnclearNoYesNoNoNoYesModerate
Juergens et al.18UnclearUnclearYes (patients and caring physicians)NoYesYesYesYesModerate
Kereiakes et al.15UnclearUnclearYes (patients and caring physicians)YesNoNoNoYesModerate
Kim et al.26UnclearUnclearNoYesNoNoYesYesModerate
Kurowski et al.27UnclearUnclearNoYesNoNoNoYesModerate
MULTISTRATEGY10Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesLow
NAPLES34UnclearUnclearNoYesNoNoNoYesModerate
Neumann et al.40UnclearUnclearNoYesNoNoNoYesModerate
Okmen et al.21UnclearUnclearNoUnclearNoNoYesYesModerate
Okmen et al.24UnclearUnclearNoYesNoNoNoYesModerate
On-TIME 2 open-label study33Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
On-TIME 233Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
Ozkan et al.28UnclearUnclearNoYesNoNoNoYesModerate
PRISM8UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
PRISM-PLUS7UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
RESTORE6UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
SASTRE23UnclearYesNoYesYesYesYesYesModerate
Shen et al.32UnclearYesNoYesYesYesYesYesModerate
STRATEGY41Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesModerate
TARGET9UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TENACITY43UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TETAMI19,20UnclearUnclearYes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TOPSTAR17UnclearYes (external personnel)Yes (patients and caring physicians)YesYesNoNoYesModerate
StudyAdequate sequence generationAllocation concealment usedBlindingConcurrent therapies similarIncomplete outcome data addressedUniform and explicit outcome definitionsFree of selective outcome reportingFree of other biasOverall risk of bias
3T/2R35Yes (computer generated)Yes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
ADVANCE25Yes (computer generated)Yes (external personnel)Yes (patients, caring physicians, and outcome assessors)YesNoYesYesYesLow
Danzi et al.37UnclearUnclearNoYesYesNoNoYesModerate
ELISA 229UnclearUnclearYes (outcome assessors)NoYesYesYesYesModerate
Ercan et al.22UnclearUnclearNoNoYesYesYesYesModerate
Ernst et al.42Yes (computer generated)UnclearNoYesNoYesYesYesModerate
EVEREST36UnclearUnclearNoYesYesNoNoYesModerate
FATA38UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
Fu et al.30UnclearUnclearNoYesNoNoYesYesModerate
Ivandic et al.31UnclearUnclearNoYesNoNoNoYesModerate
Juergens et al.18UnclearUnclearYes (patients and caring physicians)NoYesYesYesYesModerate
Kereiakes et al.15UnclearUnclearYes (patients and caring physicians)YesNoNoNoYesModerate
Kim et al.26UnclearUnclearNoYesNoNoYesYesModerate
Kurowski et al.27UnclearUnclearNoYesNoNoNoYesModerate
MULTISTRATEGY10Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesLow
NAPLES34UnclearUnclearNoYesNoNoNoYesModerate
Neumann et al.40UnclearUnclearNoYesNoNoNoYesModerate
Okmen et al.21UnclearUnclearNoUnclearNoNoYesYesModerate
Okmen et al.24UnclearUnclearNoYesNoNoNoYesModerate
On-TIME 2 open-label study33Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
On-TIME 233Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
Ozkan et al.28UnclearUnclearNoYesNoNoNoYesModerate
PRISM8UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
PRISM-PLUS7UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
RESTORE6UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
SASTRE23UnclearYesNoYesYesYesYesYesModerate
Shen et al.32UnclearYesNoYesYesYesYesYesModerate
STRATEGY41Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesModerate
TARGET9UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TENACITY43UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TETAMI19,20UnclearUnclearYes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TOPSTAR17UnclearYes (external personnel)Yes (patients and caring physicians)YesYesNoNoYesModerate
Table 3

Risk of bias assessment

StudyAdequate sequence generationAllocation concealment usedBlindingConcurrent therapies similarIncomplete outcome data addressedUniform and explicit outcome definitionsFree of selective outcome reportingFree of other biasOverall risk of bias
3T/2R35Yes (computer generated)Yes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
ADVANCE25Yes (computer generated)Yes (external personnel)Yes (patients, caring physicians, and outcome assessors)YesNoYesYesYesLow
Danzi et al.37UnclearUnclearNoYesYesNoNoYesModerate
ELISA 229UnclearUnclearYes (outcome assessors)NoYesYesYesYesModerate
Ercan et al.22UnclearUnclearNoNoYesYesYesYesModerate
Ernst et al.42Yes (computer generated)UnclearNoYesNoYesYesYesModerate
EVEREST36UnclearUnclearNoYesYesNoNoYesModerate
FATA38UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
Fu et al.30UnclearUnclearNoYesNoNoYesYesModerate
Ivandic et al.31UnclearUnclearNoYesNoNoNoYesModerate
Juergens et al.18UnclearUnclearYes (patients and caring physicians)NoYesYesYesYesModerate
Kereiakes et al.15UnclearUnclearYes (patients and caring physicians)YesNoNoNoYesModerate
Kim et al.26UnclearUnclearNoYesNoNoYesYesModerate
Kurowski et al.27UnclearUnclearNoYesNoNoNoYesModerate
MULTISTRATEGY10Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesLow
NAPLES34UnclearUnclearNoYesNoNoNoYesModerate
Neumann et al.40UnclearUnclearNoYesNoNoNoYesModerate
Okmen et al.21UnclearUnclearNoUnclearNoNoYesYesModerate
Okmen et al.24UnclearUnclearNoYesNoNoNoYesModerate
On-TIME 2 open-label study33Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
On-TIME 233Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
Ozkan et al.28UnclearUnclearNoYesNoNoNoYesModerate
PRISM8UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
PRISM-PLUS7UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
RESTORE6UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
SASTRE23UnclearYesNoYesYesYesYesYesModerate
Shen et al.32UnclearYesNoYesYesYesYesYesModerate
STRATEGY41Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesModerate
TARGET9UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TENACITY43UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TETAMI19,20UnclearUnclearYes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TOPSTAR17UnclearYes (external personnel)Yes (patients and caring physicians)YesYesNoNoYesModerate
StudyAdequate sequence generationAllocation concealment usedBlindingConcurrent therapies similarIncomplete outcome data addressedUniform and explicit outcome definitionsFree of selective outcome reportingFree of other biasOverall risk of bias
3T/2R35Yes (computer generated)Yes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
ADVANCE25Yes (computer generated)Yes (external personnel)Yes (patients, caring physicians, and outcome assessors)YesNoYesYesYesLow
Danzi et al.37UnclearUnclearNoYesYesNoNoYesModerate
ELISA 229UnclearUnclearYes (outcome assessors)NoYesYesYesYesModerate
Ercan et al.22UnclearUnclearNoNoYesYesYesYesModerate
Ernst et al.42Yes (computer generated)UnclearNoYesNoYesYesYesModerate
EVEREST36UnclearUnclearNoYesYesNoNoYesModerate
FATA38UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
Fu et al.30UnclearUnclearNoYesNoNoYesYesModerate
Ivandic et al.31UnclearUnclearNoYesNoNoNoYesModerate
Juergens et al.18UnclearUnclearYes (patients and caring physicians)NoYesYesYesYesModerate
Kereiakes et al.15UnclearUnclearYes (patients and caring physicians)YesNoNoNoYesModerate
Kim et al.26UnclearUnclearNoYesNoNoYesYesModerate
Kurowski et al.27UnclearUnclearNoYesNoNoNoYesModerate
MULTISTRATEGY10Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesLow
NAPLES34UnclearUnclearNoYesNoNoNoYesModerate
Neumann et al.40UnclearUnclearNoYesNoNoNoYesModerate
Okmen et al.21UnclearUnclearNoUnclearNoNoYesYesModerate
Okmen et al.24UnclearUnclearNoYesNoNoNoYesModerate
On-TIME 2 open-label study33Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
On-TIME 233Yes (computer generated)Yes (centralized system)Yes (outcome assessors)YesYesYesYesYesLow
Ozkan et al.28UnclearUnclearNoYesNoNoNoYesModerate
PRISM8UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
PRISM-PLUS7UnclearYes (sealed envelopes)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
RESTORE6UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
SASTRE23UnclearYesNoYesYesYesYesYesModerate
Shen et al.32UnclearYesNoYesYesYesYesYesModerate
STRATEGY41Yes (computer generated)Yes (sealed envelopes)Yes (outcome assessors)YesYesYesYesYesModerate
TARGET9UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TENACITY43UnclearYes (centralized system)Yes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TETAMI19,20UnclearUnclearYes (patients, caring physicians, and outcome assessors)YesYesYesYesYesLow
TOPSTAR17UnclearYes (external personnel)Yes (patients and caring physicians)YesYesNoNoYesModerate

Quantitative synthesis

Tirofiban vs. placebo or bivalirudin

Overall pooled effect estimate analysis showed a significant reduction in short-term (30 days) mortality [OR = 0.68 (0.54–0.86), P = 0.001, P for heterogeneity = 0.95, I2= 0%] (Figure 2), mortality or MI [OR = 0.69 (0.58–0.81), P < 0.001, P for heterogeneity=0.33, I2 = 10%] (Figure 3), MI alone [OR = 0.71 (0.56–0.90), P = 0.004, P for heterogeneity=0.16, I2 = 24%], and the composite of death, MI, or target vessel revascularization [OR = 0.73 (0.60–0.89), P = 0.002, P for heterogeneity = 0.06, I2 = 37%] in patients randomly allocated to receive tirofiban at the different bolus and infusion regimens. According to an absolute risk reduction of 2.5%, the NNT is 40 to prevent one death or MI at 30 days, with an NNT of 100 to prevent one death. Comprehensive heterogeneity and inconsistency analyses showed that included trials led to apparently statistically heterogeneous results in terms of MACE rates (P for heterogeneity = 0.06, I2 = 37%), which is paralleled by the disparities in clinical setting, tirofiban bolus and infusion regimens, interventions, duration of treatment, and outcomes. However, there was no signal of heterogeneity across trials for the composite of death or MI, mortality, or MI alone.

Figure 2

Forest plot of comparison: tirofiban vs. placebo or standard therapy, outcome: 30-day mortality rate. CI, confidence interval; Weight, statistical weight (an indirect estimate of study precision and impact on overall pooled estimates of the single study result).

Figure 3

Forest plot of comparison: tirofiban vs. placebo or standard therapy, outcome: 30-day death or myocardial infarction. CI, confidence interval; Weight, statistical weight (an indirect estimate of study precision and impact on overall pooled estimates of the single study result).

To assess the effect of tirofiban when added to P2Y12 receptor blockers (i.e. ticlopidine or clopidogrel), studies where patients were adequately pretreated with clopidogrel (n = 13) or ticlopidine (n = 1) were selected, comprising 3424 patients.17,22,24,25,27–35,42 Consistent to previous analysis, tirofiban was associated with a significant decrease in mortality [OR = 0.56 (0.34, 0.93), P = 0.02, P for heterogeneity = 0.93, I2 = 0%] and the composite of death or MI [OR = 0.61 (0.48, 0.79), P < 0.001, P for heterogeneity = 0.62, I2 = 0%] at 30 days.

The use of tirofiban tended to increase the rate of major bleeding [1.5 vs. 1.8%; OR = 1.21 (0.88, 1.67), P = 0.24, P for heterogeneity = 0.97, I2 = 0%] with an estimated NNH of 286 for one major haemorrhagic event. Minor bleedings [OR = 1.42 (1.13, 1.79), P = 0.002, P for heterogeneity = 0.97, I2 = 0%, 3.8 vs. 2.7%; NNH: 91] and the incidence of any thrombocytopenia [OR = 1.51 (1.06, 2.16); P = 0.02, P for heterogeneity = 0.96, I2 = 0%] were both significantly increased by the use of tirofiban.

After an average of 5-month follow-up, tirofiban remained associated with a significant reduction in mortality [OR = 0.81 (0.66, 0.99), P = 0.04, P = 0.70 for heterogeneity, I2 = 0%] and death or MI [OR = 0.73 (0.62, 0.85), P < 0.001, P = 0.24 for heterogeneity, I2 = 16%].

Tirofiban vs. abciximab

The overall pooled effect estimate analysis showed that tirofiban at 30 days led to similar mortality rate [OR = 0.90 (0.53, 1.54), P = 0.70, P for heterogeneity = 0.62, I2 = 0%] (Figure 4) but tended to increase the composite of death or MI [6.0 vs. 5.1%; OR = 1.18 (0.96, 1.45), P = 0.11, P for heterogeneity = 0.58, I2 = 0%] (Figure 5) and MACE rate [6.3 vs. 5.5%; OR = 1.18 (0.97, 1.44), P = 0.10, P for heterogeneity = 0.43, I2 = 0%] when compared with abciximab. Although there was no formal signal of heterogeneity across included studies, likely due to limited statistical power, these results mainly mirrored the findings of the TARGET study (study weight 78.4%) which tested tirofiban at 10 µg/kg 3 min bolus regimen and 0.15 µg/kg/min infusion. Indeed, mortality [OR = 0.73 (0.36, 1.47), P = 0.38, P for heterogeneity = 0.61, I2 = 0%], the composite of death or MI [OR = 0.87 (0.56, 1.35), P = 0.54, P for heterogeneity = 0.58, I2 = 0%] or MACE rate [OR = 0.87 (0.57, 1.32), P = 0.51, P for heterogeneity = 0.63, I2 = 0%] were similar when tirofiban at high-dose bolus was compared with abciximab.

Figure 4

Forest plot of comparison: tirofiban vs. abciximab, outcome: 30-day death. CI, confidence interval; Weight, statistical weight (an indirect estimate of study precision and impact on overall pooled estimates of the single study result).

Figure 5

Forest plot of comparison: tirofiban vs. abciximab, outcome: 30-day death or myocardial infarction. CI, confidence interval; Weight, statistical weight (an indirect estimate of study precision and impact on overall pooled estimates of the single study result).

The rate of major bleedings did not differ in tirofiban- vs. abciximab-treated patients [OR = 1.24 (0.78, 1.98), P = 0.35, P for heterogeneity = 0.76, I2 = 0%], whereas minor bleedings [3.1 vs. 4.8%; OR = 0.64 (0.50, 0.82), P < 0.001, P for heterogeneity = 0.95, I2 = 0%] and any thrombocytopenia [0.3 vs. 2.4%; OR = 0.28 (0.08, 0.94), P = 0.04, P for heterogeneity=0.71, I2 = 0%] were both markedly reduced in the tirofiban group.

At the longest available follow-up, death or MI [OR = 1.09 (0.91, 1.31), P = 0.34, P for heterogeneity = 0.87, I2 = 0%], mortality [OR = 1.03 (0.75, 1.42), P = 0.86, P for heterogeneity = 0.71, I2 = 0%], and MACE [OR = 1.00 (0.87, 1.16), P = 0.95, P for heterogeneity = 0.68, I2 = 0%] rates also did not differ between groups (Figure 6).

Figure 6

Forest plot of comparison: tirofiban vs. abciximab, outcome: long-term death or myocardial infarction. CI, confidence interval. Weight, statistical weight (an indirect estimate of study precision and impact on overall pooled estimates of the single study result).

Additional analyses

Meta-regression was performed to explore moderators of effect estimates for tirofiban vs. abciximab analysis, focusing on the 30 days rate of death or MI, and on the long-term rate of MACE, and appraising type of control, type of administration, concomitant medical treatment, adequate randomization method, adequate concealment of allocation, and adequate patient blinding (Table 4). The only potentially relevant finding beyond type of control treatment (i.e. placebo or anticoagulant vs. abciximab) was a trend for interaction between 30 days death or MI rates and the comparison between tirofiban and abciximab when focusing on the bolus regimen of tirofiban [β = −0.646 (−1.244, 0.103), P = 0.083], suggestive of more efficacy for the latter when employed at a high (25 µg/kg) dose.

Table 4

Results of meta-regression analysis

Variable30 Days death or myocardial infarction
Long-term major adverse cardiac events
Beta (95% confidence interval)P-valueBeta (95% confidence interval)P-value
Type of control0.469 (0.127; 0.767)0.0080.350 (−0.005; 0.574)0.054
Type of administration0.056 (−0.136; 0.183)0.7650.035 (−0.123; 0.149)0.850
Concomitant medical treatment0.166 (−0.190; 0.494)0.371−0.052 (−0.336; 0.255)0.782
Adequate randomization method−0.220 (−0.887; 0.226)0.234−0.203 (−0.639; 0.187)0.273
Adequate concealment of allocation0.261 (−0.099; 0.587)0.1570.035 (−0.280; 0.337)0.850
Adequate patient blinding0.362 (0.007; 0.654)0.0460.197 (−0.156; 0.507)0.289
Variable30 Days death or myocardial infarction
Long-term major adverse cardiac events
Beta (95% confidence interval)P-valueBeta (95% confidence interval)P-value
Type of control0.469 (0.127; 0.767)0.0080.350 (−0.005; 0.574)0.054
Type of administration0.056 (−0.136; 0.183)0.7650.035 (−0.123; 0.149)0.850
Concomitant medical treatment0.166 (−0.190; 0.494)0.371−0.052 (−0.336; 0.255)0.782
Adequate randomization method−0.220 (−0.887; 0.226)0.234−0.203 (−0.639; 0.187)0.273
Adequate concealment of allocation0.261 (−0.099; 0.587)0.1570.035 (−0.280; 0.337)0.850
Adequate patient blinding0.362 (0.007; 0.654)0.0460.197 (−0.156; 0.507)0.289

Based on a univariate fixed-effect model with least-squares weights for sample size to explore moderators and/or predictors of changes in log-transformed odds ratios.

Table 4

Results of meta-regression analysis

Variable30 Days death or myocardial infarction
Long-term major adverse cardiac events
Beta (95% confidence interval)P-valueBeta (95% confidence interval)P-value
Type of control0.469 (0.127; 0.767)0.0080.350 (−0.005; 0.574)0.054
Type of administration0.056 (−0.136; 0.183)0.7650.035 (−0.123; 0.149)0.850
Concomitant medical treatment0.166 (−0.190; 0.494)0.371−0.052 (−0.336; 0.255)0.782
Adequate randomization method−0.220 (−0.887; 0.226)0.234−0.203 (−0.639; 0.187)0.273
Adequate concealment of allocation0.261 (−0.099; 0.587)0.1570.035 (−0.280; 0.337)0.850
Adequate patient blinding0.362 (0.007; 0.654)0.0460.197 (−0.156; 0.507)0.289
Variable30 Days death or myocardial infarction
Long-term major adverse cardiac events
Beta (95% confidence interval)P-valueBeta (95% confidence interval)P-value
Type of control0.469 (0.127; 0.767)0.0080.350 (−0.005; 0.574)0.054
Type of administration0.056 (−0.136; 0.183)0.7650.035 (−0.123; 0.149)0.850
Concomitant medical treatment0.166 (−0.190; 0.494)0.371−0.052 (−0.336; 0.255)0.782
Adequate randomization method−0.220 (−0.887; 0.226)0.234−0.203 (−0.639; 0.187)0.273
Adequate concealment of allocation0.261 (−0.099; 0.587)0.1570.035 (−0.280; 0.337)0.850
Adequate patient blinding0.362 (0.007; 0.654)0.0460.197 (−0.156; 0.507)0.289

Based on a univariate fixed-effect model with least-squares weights for sample size to explore moderators and/or predictors of changes in log-transformed odds ratios.

Inspection of funnel plots for either tirofiban vs. control and tirofiban vs. abciximab and the 30 days rate of death or MI (Figures 7 and 8) did not disclose evidence of small study bias, which was also confirmed by analytical testing with Peters test (P = 0.365 and P = 0.921, respectively).

Figure 7

Funnel plot for the long-term risk of major adverse cardiac events (MACE) comparing tirofiban vs. control. This plot shows the association (or lack of) between study effect (x-axis) and study size/precision (y-axis), and can thus provide a graphical appraisal of the risk of small study bias in the overall systematic review. Specifically, small study bias, also known as publication bias, is due to the selective reporting and publication of small but significant studies and the selective under-reporting and lack of publication of small non-significant studies. If present, small study bias may unduly impact on pooled effect estimates and bias the overall results toward rejecting a null hypothesis which is actually valid. The vertical dashed line represents the summary pooled effect estimate, the oblique dashed lines represent the corresponding 95% confidence intervals, and the P-value provided by analytical testing with Peters test. OR, odds ratio; SE, standard error.

Figure 8

Funnel plot for the long-term risk of major adverse cardiac events (MACE) comparing tirofiban vs. abciximab. This plot shows the association (or lack of) between study effect (x-axis) and study size/precision (y-axis), and can thus provide a graphical appraisal of the risk of small study bias in the overall systematic review. Specifically, small study bias, also known as publication bias, is due to the selective reporting and publication of small but significant studies and the selective under-reporting and lack of publication of small non-significant studies. If present, small study bias may unduly impact on pooled effect estimates and bias the overall results toward rejecting a null hypothesis which is actually valid. The vertical dashed line represents the summary pooled effect estimate, the oblique dashed lines represent the corresponding 95% confidence intervals, and the P-value provided by analytical testing with Peters test. OR, odds ratio; SE, standard error.

Discussion

The main finding of this meta-analysis is that adjunctive tirofiban therapy, compared with placebo, is associated with a >30% reduction in all considered ischaemic endpoints including overall mortality, mortality or MI, and MACE rates within 30 days after treatment. In absolute terms, tirofiban administration in 40 patients would prevent one death or MI, whereas 100 treated patients would lead to one fatal event prevention. Importantly, the benefit observed soon after intervention persisted at longest available follow-up. Interestingly, the magnitude of benefit for mortality observed in our analysis for tirofiban was quite similar to the treatment benefit shown by abciximab in a recent meta-analysis.44

As expected, the advantage in terms of ischaemic endpoints was counterbalanced by a significant increase in minor, but not major, bleeding and thrombocytopenia. Assuming that the observed insignificant 25% relative increase in major bleeding in the tirofiban group is real, we estimated a NNH of 286, 91, and 227 to lead to one major bleed, one minor haemorrhagic event, and one episode of thrombocytopenia, respectively. Thus, altogether the use of tirofiban at different tested regimens was associated with a favourable efficacy/safety profile in a broad patient population presenting with acute coronary syndromes and/or undergoing PCI.

While several included studies antedated the advent of clopidogrel pre-treatment strategy in patients undergoing PCI, our sensitivity analysis, which focused on patients receiving tirofiban on top of pretreatment with clopidogrel or ticlopidine,17,22,24,25,27–35,42 suggested the benefit of tirofiban to be additive to first or second generation P2Y12 receptor inhibitors. These findings are in keeping with previous evidence45,46 and reinforce the importance of the degree and consistency of platelet inhibition to prevent ischaemic complications in patients with acute coronary syndromes undergoing PCI.

Our analysis failed to show heterogeneity of results across the different tested regimens of tirofiban for mortality or the composite of death or MI. However, for both MI rate alone and the composite of MACE rate, some degree of inconsistency was noted throughout. This might be due to various MI definitions, multiple clinical settings and/or different tirofiban tested regimens throughout studies. Interestingly, trials testing tirofiban downstream at high bolus dose, which results in a prompt and significantly greater inhibition of platelet activity compared with both standard 10 µg/kg 3 min and 0.4 µg/kg 30 min bolus regimens,3,47,48 resulted in overall numerically higher relative and absolute reduction of death or MI, MI alone, and MACE rates within the first 30 days.

In aggregate, ischaemic complications did not significantly differ in tirofiban vs. abciximab-treated patients at short- or medium-term follow-up. However, tirofiban tested at 10 µg/kg bolus regimen, which results in suboptimal platelet inhibition soon after administration,3,47 increased peri-procedural ischaemic events mainly in terms of MI, compared with abciximab. This was largely driven by the results of the TARGET study, which remains by far the biggest comparison between the two drugs.9 In contrast, the 25 µg/kg tirofiban bolus regimen which has been developed to more closely mimic abciximab-driven platelet inhibition soon after treatment administration,48 was not associated with an increase of early ischaemic hazard when contrasted to the latter. Indeed, a trend was noted suggesting an interaction between 30-day death or MI rates and the comparison between tirofiban and abciximab when focusing on dosage of tirofiban administration. While abciximab treatment effect vs. placebo was previously shown to be directly proportional to risk status of treated patients,49 no such pattern was observed when abciximab was compared with tirofiban at metaregression analysis, suggesting that tirofiban may effectively replace abciximab across the whole spectrum of patients with CAD, particularly with a high-dose bolus regimen.

Altogether, the pooled findings from both placebo and abciximab controlled studies suggest that the bolus regimen, especially for patients undergoing PCI and receiving treatment immediately before is of utmost importance to optimize outcomes. Tirofiban, given at a high-dose bolus, by providing a greater and more consistent level of platelet inhibition may be a preferable option than previously developed standard regimens which lead to desirable anti-platelet activity only with some delay after drug administration.47

Importantly, confidence intervals around point of estimate for ischaemic events remains wide for the comparison between tirofiban and abciximab and entail the possibility that even at high bolus regimen, the former may lead to a relatively small yet distinct increase in adverse events after PCI. This uncertainty largely reflects the still limited number of patients who have been re-evaluated in head-to-head studies with tirofiban given at high bolus dose. Unfortunately, the planned large (n = 8800 patients) TENACITY study which aimed to definitively ascertain whether at proper dosing tirofiban would be non-inferior to abciximab was prematurely stopped for financial reasons after 383 patients were enrolled.43 All subsequent investigator-driven head-to-head comparisons between these two agents were based on surrogate endpoints such as ST-segment elevation resolution,10,38,41 myocardial blush,36 left ventricular ejection fraction,37 or platelet inhibition42 which explains the relatively small study populations.

An additional finding of potential clinical relevance was that the rate of minor, but not major, bleeding was significantly reduced by the use of tirofiban compared with abciximab. This was consistently noted in studies testing either 10 or 25 µg/kg tirofiban bolus regimens. Since the degree of platelet inhibition provided by a high-dose tirofiban bolus is not inferior to that of abciximab, and indeed many previous studies have shown that tirofiban at this revised bolus regimen might be associated with greater and more consistent anti-platelet activity than abciximab,41,42,50 this observation of lower minor bleeding rate in tirofiban-treated patients is intriguing and deserves further investigation. Similarly, the rate of thrombocytopenia, which like bleeding complications has been shown to independently predict worse outcomes,51,52 was reduced by almost 80% by the use of tirofiban. This likely reflects the lower propensity of tirofiban to elicit an antibody response. Thrombocytopenia has been shown to be associated with bleeding complications,51,52 and it is tempting to speculate that the lower propensity of tirofiban to trigger an immune response might at least partially explain the improved safety profile in terms of minor bleedings observed in the tirofiban group. Finally, we cannot rule out the possibility that the difference in minor bleedings noted between tirofiban and abciximab is a spurious finding or simply related to the shorter duration of anti-aggregatory effect.

Study limitations

Our results suffer from those limitations which are inherent to all meta-analytic techniques including particularly heterogeneity in patient populations, different study drug regimens, and variable endpoint definitions across studies. This mainly applies to the different criteria employed throughout trials for classifying bleeding and peri-procedural ischaemic endpoints. Importantly, however, a clear reduction of overall mortality in the tirofiban arm has been noted vs. placebo but not vs. abciximab studies which is in keeping with the differences observed between study groups for MI alone or the composite of death or MI.

Conclusions

In our pooled analysis based on over 20 000 patients, tirofiban administration was shown to significantly reduce mortality, the composite of death or MI along with MACE rate when compared with placebo. This benefit in ischaemic endpoints reduction remained significant and of consistent magnitude in studies where tirofiban was tested in addition to thienopyridines but came at an increase risk for minor bleeding and thrombocytopenia. An early ischaemic hazard disfavouring tirofiban was noted when compared with abciximab in studies based on 10 µg/kg bolus regimen but not in those testing the 25 µg/kg bolus regimen. Overall, the safety profile seems to favour the use of tirofiban over abciximab for lower incidence of minor bleeding and thrombocytopenia, likely reflecting different chemical structures more than a difference in anti-platelet potency between these two drugs.

Our findings suggest that the use of tirofiban is an efficacious treatment option to reduce ischaemic events in patients with acute coronary syndromes and/or those undergoing PCI. When employed at high-dose bolus just prior to PCI, tirofiban may provide similar efficacy yet an improved safety profile when compared with abciximab. This hypothesis would require prospective assessment in order to be validated.

Funding

This work was supported by the University of Ferrara, Italy.

Conflicts of interest: M.V. has consulted for Iroko, Eli Lilly and the Medicines Company, has lectured for Iroko, Glaxo SmithKline and received grant support from Iroko and Eli Lilly. G.B.-Z. has consulted for Cordis and The Medicines Company, has lectured for Bristol-Myers Squibb and sanofi-aventis, and has received grant support from Glaxo SmithKline.

References

1
Scarborough
RM
Kleiman
NS
Phillips
DR
Platelet glycoprotein IIb/IIIa antagonists. What are the relevant issues concerning their pharmacology and clinical use?
Circulation
1999
, vol. 
100
 (pg. 
437
-
444
)
2
Topol
EJ
Byzova
TV
Plow
EF
Platelet GPIIb-IIIa blockers
Lancet
1999
, vol. 
353
 (pg. 
227
-
231
)
3
Batchelor
WB
Tolleson
TR
Huang
Y
Larsen
RL
Mantell
RM
Dillard
P
Davidian
M
Zhang
D
Cantor
WJ
Sketch
MH
Jr
Ohman
EM
Zidar
JP
Gretler
D
DiBattiste
PM
Tcheng
JE
Califf
RM
Harrington
RA
Randomized COMparison of platelet inhibition with abciximab, tiRofiban and eptifibatide during percutaneous coronary intervention in acute coronary syndromes: the COMPARE trial. Comparison of Measurements of Platelet aggregation with Aggrastat, Reopro, and Eptifibatide
Circulation
2002
, vol. 
106
 (pg. 
1470
-
1476
)
4
Lele
M
Sajid
M
Wajih
N
Stouffer
GA
Eptifibatide and 7E3, but not tirofiban, inhibit alpha(v)beta(3) integrin-mediated binding of smooth muscle cells to thrombospondin and prothrombin
Circulation
2001
, vol. 
104
 (pg. 
582
-
587
)
5
Reininger
AJ
Agneskirchner
J
Bode
PA
Spannagl
M
Wurzinger
LJ
c7E3 Fab inhibits low shear flow modulated platelet adhesion to endothelium and surface-absorbed fibrinogen by blocking platelet GP IIb/IIIa as well as endothelial vitronectin receptor—results from patients with acute myocardial infarction and healthy controls
Thromb Haemost
2000
, vol. 
83
 (pg. 
217
-
223
)
6
The RESTORE Investigators
Effects of platelet glycoprotein IIb/IIIa blockade with tirofiban on adverse cardiac events in patients with unstable angina or acute myocardial infarction undergoing coronary angioplasty. Randomized Efficacy Study of Tirofiban for Outcomes and REstenosis
Circulation
1997
, vol. 
96
 (pg. 
1445
-
1453
)
7
Platelet Receptor Inhibition in Ischemic Syndrome Management in Patients Limited by Unstable Signs and Symptoms (PRISM-PLUS) Study Investigators
Inhibition of the platelet glycoprotein IIb/IIIa receptor with tirofiban in unstable angina and non-Q-wave myocardial infarction
N Engl J Med
1998
, vol. 
338
 (pg. 
1488
-
1497
)
8
Platelet Receptor Inhibition in Ischemic Syndrome Management (PRISM) Study Investigators
A comparison of aspirin plus tirofiban with aspirin plus heparin for unstable angina
N Engl J Med
1998
, vol. 
338
 (pg. 
1498
-
1505
)
9
Topol
EJ
Moliterno
DJ
Herrmann
HC
Powers
ER
Grines
CL
Cohen
DJ
Cohen
EA
Bertrand
M
Neumann
FJ
Stone
GW
DiBattiste
PM
Demopoulos
L
Comparison of two platelet glycoprotein IIb/IIIa inhibitors, tirofiban and abciximab, for the prevention of ischemic events with percutaneous coronary revascularization
N Engl J Med
2001
, vol. 
344
 (pg. 
1888
-
1894
)
10
Valgimigli
M
Campo
G
Percoco
G
Bolognese
L
Vassanelli
C
Colangelo
S
de Cesare
N
Rodriguez
AE
Ferrario
M
Moreno
R
Piva
T
Sheiban
I
Pasquetto
G
Prati
F
Nazzaro
MS
Parrinello
G
Ferrari
R
Comparison of angioplasty with infusion of tirofiban or abciximab and with implantation of sirolimus-eluting or uncoated stents for acute myocardial infarction: the MULTISTRATEGY randomized trial
J Am Med Assoc
2008
, vol. 
299
 (pg. 
1788
-
1799
)
11
Higgins
JPT
Green
SG
Cochrane Handbook for Systematic Reviews of Interventions
2008
 
Version 501. Available from http://www.cochrane-handbookorg (last accessed on 18 May 2009)
12
Wong
SS
Wilczynski
NL
Haynes
RB
Comparison of top-performing search strategies for detecting clinically sound treatment studies and systematic reviews in MEDLINE and EMBASE
J Med Libr Assoc
2006
, vol. 
94
 (pg. 
451
-
455
)
13
TIMI Study Group
The Thrombolysis in Myocardial Infarction (TIMI) trial. Phase I findings
N Engl J Med
1985
, vol. 
312
 (pg. 
932
-
936
)
14
Peters
JL
Sutton
AJ
Jones
DR
Abrams
KR
Rushton
L
Comparison of two methods to detect publication bias in meta-analysis
J Am Med Assoc
2006
, vol. 
295
 (pg. 
676
-
680
)
15
Kereiakes
DJ
Kleiman
NS
Ambrose
J
Cohen
M
Rodriguez
S
Palabrica
T
Herrmann
HC
Sutton
JM
Weaver
WD
McKee
DB
Fitzpatrick
V
Sax
FL
Randomized, double-blind, placebo-controlled dose-ranging study of tirofiban (MK-383) platelet IIb/IIIa blockade in high risk patients undergoing coronary angioplasty
J Am Coll Cardiol
1996
, vol. 
27
 (pg. 
536
-
542
)
16
Gibson
CM
Goel
M
Cohen
DJ
Piana
RN
Deckelbaum
LI
Harris
KE
King
SB
III
Six-month angiographic and clinical follow-up of patients prospectively randomized to receive either tirofiban or placebo during angioplasty in the RESTORE trial. Randomized Efficacy Study of Tirofiban for Outcomes and Restenosis
J Am Coll Cardiol
1998
, vol. 
32
 (pg. 
28
-
34
)
17
Bonz
AW
Lengenfelder
B
Strotmann
J
Held
S
Turschner
O
Harre
K
Wacker
C
Waller
C
Kochsiek
N
Meesmann
M
Neyses
L
Schanzenbacher
P
Ertl
G
Voelker
W
Effect of additional temporary glycoprotein IIb/IIIa receptor inhibition on troponin release in elective percutaneous coronary interventions after pretreatment with aspirin and clopidogrel (TOPSTAR trial)
J Am Coll Cardiol
2002
, vol. 
40
 (pg. 
662
-
668
)
18
Juergens
CP
White
HD
Belardi
JA
Macaya
C
Soler-Soler
J
Meyer
BJ
Levy
RD
Bunt
T
Menten
J
Herrmann
HC
Adgey
AA
Tarnesby
G
A multicenter study of the tolerability of tirofiban versus placebo in patients undergoing planned intracoronary stent placement
Clin Ther
2002
, vol. 
24
 (pg. 
1332
-
1344
)
19
Cohen
M
Gensini
GF
Maritz
F
Gurfinkel
EP
Huber
K
Timerman
A
Krzeminska-Pakula
M
Danchin
N
White
HD
Santopinto
J
Bigonzi
F
Hecquet
C
Vittori
L
The safety and efficacy of subcutaneous enoxaparin versus intravenous unfractionated heparin and tirofiban versus placebo in the treatment of acute ST-segment elevation myocardial infarction patients ineligible for reperfusion (TETAMI): a randomized trial
J Am Coll Cardiol
2003
, vol. 
42
 (pg. 
1348
-
1356
)
20
Cohen
M
Gensini
GF
Maritz
F
Gurfinkel
EP
Huber
K
Timerman
A
Krzeminska-Pakula
M
Santopinto
J
Hecquet
C
Vittori
L
Prospective evaluation of clinical outcomes after acute ST-elevation myocardial infarction in patients who are ineligible for reperfusion therapy: preliminary results from the TETAMI registry and randomized trial
Circulation
2003
, vol. 
108
 (pg. 
III14
-
III21
)
21
Okmen
E
Cakmak
M
Tartan
Z
Cam
N
Effects of glycoprotein IIb/IIIa inhibition on clinical stabilization parameters in patients with unstable angina and non-Q-wave myocardial infarction
Heart Vessels
2003
, vol. 
18
 (pg. 
117
-
122
)
22
Ercan
E
Tengiz
I
Duman
C
Onbasili
OA
Baris
N
Effect of tirofiban on C-reactive protein in non-ST-elevation myocardial infarction
Am Heart J
2004
, vol. 
147
 pg. 
E1
 
23
Martinez-Rios
MA
Rosas
M
Gonzalez
H
Pena-Duque
MA
Martinez-Sanchez
C
Gaspar
J
Garcia
H
Gaxiola
E
Delgado
L
Carrillo
J
Leyva
JL
Lupi
E
Comparison of reperfusion regimens with or without tirofiban in ST-elevation acute myocardial infarction
Am J Cardiol
2004
, vol. 
93
 (pg. 
280
-
287
)
24
Okmen
E
Sanli
A
Uyarel
H
Dayi
S
Tartan
Z
Cam
N
Effects of glycoprotein IIb/IIIa receptor inhibition with tirofiban on minor myocardial damage in angiographically successful percutaneous coronary angioplasty
Cardiology
2004
, vol. 
102
 (pg. 
18
-
23
)
25
Valgimigli
M
Percoco
G
Barbieri
D
Ferrari
F
Guardigli
G
Parrinello
G
Soukhomovskaia
O
Ferrari
R
The additive value of tirofiban administered with the high-dose bolus in the prevention of ischemic complications during high-risk coronary angioplasty: the ADVANCE Trial
J Am Coll Cardiol
2004
, vol. 
44
 (pg. 
14
-
19
)
26
Kim
JH
Jeong
MH
Rhew
JY
Lim
JH
Yun
KH
Kim
KH
Kang
DK
Hong
SN
Lim
SY
Lee
SH
Lee
YS
Hong
YJ
Park
HW
Kim
W
Ahn
YK
Moon
Y
Cho
JG
Park
JC
Kang
JC
Long-term clinical outcomes of platelet glycoprotein IIb/IIIa inhibitor combined with low molecular weight heparin in patients with acute coronary syndrome
Circ J
2005
, vol. 
69
 (pg. 
159
-
164
)
27
Kurowski
V
Toelg
R
Jain
D
Richter
C
Wiegand
UK
Richardt
G
Khattab
AA
Effect of adjunctive treatment with tirofiban on troponin T elevation during stenting of critically stenosed aortocoronary saphenous vein grafts
Am J Cardiol
2005
, vol. 
96
 (pg. 
681
-
684
)
28
Ozkan
M
Sag
C
Yokusoglu
M
Uzun
M
Baysan
O
Erinc
K
Isik
E
The effect of tirofiban and clopidogrel pretreatment on outcome of old saphenous vein graft stenting in patients with acute coronary syndromes
Tohoku J Exp Med
2005
, vol. 
206
 (pg. 
7
-
13
)
29
Rasoul
S
Ottervanger
JP
de Boer
MJ
Miedema
K
Hoorntje
JC
Gosselink
M
Zijlstra
F
Suryapranata
H
Dambrink
JH
van ’t Hof
AW
A comparison of dual vs. triple antiplatelet therapy in patients with non-ST-segment elevation acute coronary syndrome: results of the ELISA-2 trial
Eur Heart J
2006
, vol. 
27
 (pg. 
1401
-
1407
)
30
Fu
XH
Hao
QQ
Jia
XW
Fan
WZ
Gu
XS
Wu
WL
Hao
GZ
Li
SQ
Jiang
YF
Geng
W
Effect of tirofiban plus clopidogrel and aspirin on primary percutaneous coronary intervention via transradial approach in patients with acute myocardial infarction
Chin Med J
2008
, vol. 
121
 (pg. 
522
-
527
)
31
Ivandic
BT
Kurz
K
Keck
F
Staritz
P
Lehrke
S
Katus
HA
Giannitsis
E
Tirofiban optimizes platelet inhibition for immediate percutaneous coronary intervention in high-risk acute coronary syndromes
Thromb Haemost
2008
, vol. 
100
 (pg. 
648
-
654
)
32
Shen
J
Zhang
Q
Zhang
RY
Zhang
JS
Hu
J
Yang
ZK
Zheng
AF
Zhang
X
Shen
WF
Clinical benefits of adjunctive tirofiban therapy in patients with acute ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention
Coron Artery Dis
2008
, vol. 
19
 (pg. 
271
-
277
)
33
Van 't Hof
AW
ten Berg
J
Heestermans
T
Dill
T
Funck
RC
van Werkum
W
Dambrink
JH
Suryapranata
H
van Houwelingen
G
Ottervanger
JP
Stella
P
Giannitsis
E
Hamm
C
Prehospital initiation of tirofiban in patients with ST-elevation myocardial infarction undergoing primary angioplasty (On-TIME 2): a multicentre, double-blind, randomised controlled trial
Lancet
2008
, vol. 
372
 (pg. 
537
-
546
)
34
Briguori
C
NAPLES: a prospective, randomized trial comparison of bivalirudin and unfractionated heparin plus tirofiban in patients with diabetes mellitus undergoing elective percutaneous coronary intervention
2008
 
35
Valgimigli
M
Main results of the tailoring treatment with tirofiban in patients showing resistance to aspirin or resistance to clopidogrel study (3T/2R)
2008
 
36
Bolognese
L
Falsini
G
Liistro
F
Angioli
P
Ducci
K
Taddei
T
Tarducci
R
Cosmi
F
Baldassarre
S
Burali
A
Randomized comparison of upstream tirofiban versus downstream high bolus dose tirofiban or abciximab on tissue-level perfusion and troponin release in high-risk acute coronary syndromes treated with percutaneous coronary interventions: the EVEREST trial
J Am Coll Cardiol
2006
, vol. 
47
 (pg. 
522
-
528
)
37
Danzi
GB
Sesana
M
Capuano
C
Mauri
L
Berra Centurini
P
Baglini
R
Comparison in patients having primary coronary angioplasty of abciximab versus tirofiban on recovery of left ventricular function
Am J Cardiol
2004
, vol. 
94
 (pg. 
35
-
39
)
38
Marzocchi
A
Manari
A
Piovaccari
G
Marrozzini
C
Marra
S
Magnavacchi
P
Sangiorgio
P
Marinucci
L
Taglieri
N
Gordini
G
Binetti
N
Guiducci
V
Franco
N
Reggiani
ML
Saia
F
Randomized comparison between tirofiban and abciximab to promote complete ST-resolution in primary angioplasty: results of the facilitated angioplasty with tirofiban or abciximab (FATA) in ST-elevation myocardial infarction trial
Eur Heart J
2008
, vol. 
29
 (pg. 
2972
-
2980
)
39
Mukherjee
D
Topol
EJ
Bertrand
ME
Kristensen
SD
Herrmann
HC
Neumann
FJ
Yakubov
SJ
Bassand
JP
McClure
RR
Stone
GW
Ardissino
D
Moliterno
DJ
Mortality at 1 year for the direct comparison of tirofiban and abciximab during percutaneous coronary revascularization: do tirofiban and ReoPro give similar efficacy outcomes at trial 1-year follow-up
Eur Heart J
2005
, vol. 
26
 (pg. 
2524
-
2528
)
40
Neumann
FJ
Hochholzer
W
Pogatsa-Murray
G
Schomig
A
Gawaz
M
Antiplatelet effects of abciximab, tirofiban and eptifibatide in patients undergoing coronary stenting
J Am Coll Cardiol
2001
, vol. 
37
 (pg. 
1323
-
1328
)
41
Valgimigli
M
Percoco
G
Malagutti
P
Campo
G
Ferrari
F
Barbieri
D
Cicchitelli
G
McFadden
EP
Merlini
F
Ansani
L
Guardigli
G
Bettini
A
Parrinello
G
Boersma
E
Ferrari
R
Tirofiban and sirolimus-eluting stent vs abciximab and bare-metal stent for acute myocardial infarction: a randomized trial
J Am Med Assoc
2005
, vol. 
293
 (pg. 
2109
-
2117
)
42
Ernst
NM
Suryapranata
H
Miedema
K
Slingerland
RJ
Ottervanger
JP
Hoorntje
JC
Gosselink
AT
Dambrink
JH
de Boer
MJ
Zijlstra
F
van ’t Hof
AW
Achieved platelet aggregation inhibition after different antiplatelet regimens during percutaneous coronary intervention for ST-segment elevation myocardial infarction
J Am Coll Cardiol
2004
, vol. 
44
 (pg. 
1187
-
1193
)
43
Moliterno
DJ
Tirofiban Evaluation of Novel Dosing vs. Abciximab with Clopidogrel and Inhibition of Thrombin Study. The TENACITY
2005
 
Presented at TCT
44
De Luca
G
Suryapranata
H
Stone
GW
Antoniucci
D
Tcheng
JE
Neumann
FJ
Van de Werf
F
Antman
EM
Topol
EJ
Abciximab as adjunctive therapy to reperfusion in acute ST-segment elevation myocardial infarction: a meta-analysis of randomized trials
J Am Med Assoc
2005
, vol. 
293
 (pg. 
1759
-
1765
)
45
Kastrati
A
Mehilli
J
Neumann
FJ
Dotzer
F
ten Berg
J
Bollwein
H
Graf
I
Ibrahim
M
Pache
J
Seyfarth
M
Schuhlen
H
Dirschinger
J
Berger
PB
Schomig
A
Abciximab in patients with acute coronary syndromes undergoing percutaneous coronary intervention after clopidogrel pretreatment: the ISAR-REACT 2 randomized trial
J Am Med Assoc
2006
, vol. 
295
 (pg. 
1531
-
1538
)
46
Gurbel
PA
Bliden
KP
Zaman
KA
Yoho
JA
Hayes
KM
Tantry
US
Clopidogrel loading with eptifibatide to arrest the reactivity of platelets: results of the Clopidogrel Loading With Eptifibatide to Arrest the Reactivity of Platelets (CLEAR PLATELETS) study
Circulation
2005
, vol. 
111
 (pg. 
1153
-
1159
)
47
Kabbani
SS
Aggarwal
A
Terrien
EF
DiBattiste
PM
Sobel
BE
Schneider
DJ
Suboptimal early inhibition of platelets by treatment with tirofiban and implications for coronary interventions
Am J Cardiol
2002
, vol. 
89
 (pg. 
647
-
650
)
48
Schneider
DJ
Herrmann
HC
Lakkis
N
Aguirre
F
Lo
MW
Yin
KC
Aggarwal
A
Kabbani
SS
DiBattiste
PM
Increased concentrations of tirofiban in blood and their correlation with inhibition of platelet aggregation after greater bolus doses of tirofiban
Am J Cardiol
2003
, vol. 
91
 (pg. 
334
-
336
)
49
De Luca
G
Suryapranata
H
Stone
GW
Antoniucci
D
Tcheng
JE
Neumann
FJ
Bonizzoni
E
Topol
EJ
Chiariello
M
Relationship between patient's risk profile and benefits in mortality from adjunctive abciximab to mechanical revascularization for ST-segment elevation myocardial infarction: a meta-regression analysis of randomized trials
J Am Coll Cardiol
2006
, vol. 
47
 (pg. 
685
-
686
)
50
Danzi
GB
Capuano
C
Sesana
M
Mauri
L
Sozzi
FB
Variability in extent of platelet function inhibition after administration of optimal dose of glycoprotein IIb/IIIa receptor blockers in patients undergoing a high-risk percutaneous coronary intervention
Am J Cardiol
2006
, vol. 
97
 (pg. 
489
-
493
)
51
Merlini
PA
Rossi
M
Menozzi
A
Buratti
S
Brennan
DM
Moliterno
DJ
Topol
EJ
Ardissino
D
Thrombocytopenia caused by abciximab or tirofiban and its association with clinical outcome in patients undergoing coronary stenting
Circulation
2004
, vol. 
109
 (pg. 
2203
-
2206
)
52
McClure
MW
Berkowitz
SD
Sparapani
R
Tuttle
R
Kleiman
NS
Berdan
LG
Lincoff
AM
Deckers
J
Diaz
R
Karsch
KR
Gretler
D
Kitt
M
Simoons
M
Topol
EJ
Califf
RM
Harrington
RA
Clinical significance of thrombocytopenia during a non-ST-elevation acute coronary syndrome. The platelet glycoprotein IIb/IIIa in unstable angina: receptor suppression using integrilin therapy (PURSUIT) trial experience
Circulation
1999
, vol. 
99
 (pg. 
2892
-
2900
)

Supplementary data